HUE030838T2 - Recombinant human cc10 protein for treatment of influenza - Google Patents

Recombinant human cc10 protein for treatment of influenza Download PDF

Info

Publication number
HUE030838T2
HUE030838T2 HUE10824032A HUE10824032A HUE030838T2 HU E030838 T2 HUE030838 T2 HU E030838T2 HU E10824032 A HUE10824032 A HU E10824032A HU E10824032 A HUE10824032 A HU E10824032A HU E030838 T2 HUE030838 T2 HU E030838T2
Authority
HU
Hungary
Prior art keywords
uteroglobin
ahol ahol
human
influenza
rhcc10
Prior art date
Application number
HUE10824032A
Other languages
Hungarian (hu)
Inventor
Aprile L Pilon
Pierre Borgeat Ph D
Louis Flamand Ph D
Original Assignee
Therabron Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Therabron Therapeutics Inc filed Critical Therabron Therapeutics Inc
Publication of HUE030838T2 publication Critical patent/HUE030838T2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16111Influenzavirus A, i.e. influenza A virus
    • C12N2760/16122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16111Influenzavirus A, i.e. influenza A virus
    • C12N2760/16134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Virology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Molecular Biology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pulmonology (AREA)
  • Biochemistry (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Otolaryngology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)
  • Peptides Or Proteins (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Description

Description
CROSS REFERENCE TO RELATED APPLICATIONS
[0001] This application is a PCT International Application which daims the benefit of U .S. Provisional Application No. 61/252,028, filed October 15, 2009.
FIELD OF THE INVENTION
[0002] There are disclosed methods of reducing viral titers in vivo and treating a viral respiratory infection in a patient, methods of treating influenza infection, including Type A influenza, particularly H1N1 influenza, and methods of treating the above using intranasally-administered and/or intravenously-administered, and/or inhaled recombinant human CC10.
BACKGROUND
[0003] Clara Cell "10 kDa" protein (CC10) or uteroglobin (UG) is a small, homodimeric secretory protein produced by several mucosal epithelia and other organs of epithelial origin (Mukherjee, 1999). CC10 consists of two identical subunits of 70 amino acid residues, each with the "four helical bundle" secondary structure motif, joined in antiparallel orientation by two disulfide bonds between Cys 3 and 69’, 3’ and 69 (Matthews, 1994; Morize, 1997). CC10 is the first member of an emerging family of small globular proteins that share the same secondary, tertiary and quaternary structure and are thought to mediate similar functions. The homodimer containing two disulfide bonds appears to be its primary form. In humans, the lung is the main site of CC10 production, while several other organs synthesize smaller amounts of mRNA encoding this protein (Singh, 1987; Sandmoller, 1994). CC10 is an anti-inflammatory and immunomodulatory protein that has been characterized with respect to various interactions with other proteins, receptors and cell types (reviewed in Mukherjee, 1999 and Pilon, 2000). Lower levels of CC10 protein or mRNA have been found in various tissue and fluid samples for a number of clinical conditions characterized by some degree of inflammation including pneumonia (Nomori, 1995).
[0004] The physiology of CC10 protein in different types of pulmonary infections has been studied in one strain of CC10 knockout mouse. In two studies in which CC10 knockout and wild type mice were each infected with either Pseudomonas aeruginosa or adenovirus, two common human respiratory pathogens, the wild type mice experienced more rapid clearing of the pathogens, with greater killing of the pathogens by the innate immune system, suggesting a benefit to CC10 deficiency during viral and bacterial infection (Hayashida, 1999; Harrod, 1998). This is consistent with earlier observations in which CC10 was reported to be an immunosuppressive agent (Dierynck, 1995; 1996), indicating that CC10 would suppress the natural immune response to an infection, whether bacterial or viral, including influenza. Thus, the administration of CC10 in the presence of a viral or bacterial respiratory infection could not be expected to benefit the patient. Subsequently, it was reported that restoration of CC10 function using recombinant human CC10 protein (rhCC10) prior to infection with respiratory syncytial virus (RSV), enabled a more rapid clearance of the infection than in untreated knockout mice (Wang, 2003). However, a recent study showed that rhCC10 can pre-1 vent the development of acquired immunity, specifically antigen-specific T cells, when present at the same time that dendritic cells are exposed to antigen (Johansson, 2007), which again indicates that administration of rhCC10 may not benefit a patient with an infection. Thus, the current state of knowledge regarding the potential hazards or benefits of rhCC10 treatment during a respiratory infection is conflicting and allows no conclusions to be drawn regarding the safe and/or efficacious use of CC10 to treat different types of respiratory infections. No 1 information regarding the effect of CC10 on influenza infection is available. We report herein, direct verification of the efficacy of rhCC10 against influenza Type A in vivo, direct verification of the anti-viral effects of CC10 at the cellular level, its mechanism of action, and its potential use to treat and/or prevent viral infection, and, in particular, influenza infection.
[0005] Influenza has caused four major outbreaks (1889,1918,1957, and 1968) in the past 120 years, causing the deaths of an estimated 50-100 million people 1 worldwide. Influenza is an orthomyxovirus, an RNA virus that is transmitted by aerosols as well as by direct contact of contaminated surfaces with nasal mucosa and targets respiratory epithelial cells. Influenza infection may cause severe symptoms, including fever, sore throat and mus-1 cle aches, malaise, weight loss, respiratory congestion, and sometimes respiratory failure and death. Influenza elicits an acquired immune response (cytotoxic T cells and antibodies) that typically clears the infection in 1-2 weeks in normal healthy individuals. Several subtypes 1 of influenza that infect humans, including avian influenza (H5N1), seasonal influenza H3N2, and swine flu (H1N1), can be treated with antiviral agents such as neuraminidase inhibitors. However, the rapid rate of mutation in influenza has led to the development of drug-resistant ' strains (Moscona, 2009), such that widespread use of antiviral agents for prevention and or treatment will lead to acceleration of the development of resistance to these drugs. New therapeutic agents are therefore needed to treat, cure and prevent influenza infection. Likewise, 1 there are no approved therapies for the vast majority of viral infections in the respiratory tract and other body systems.
OBJECTS OF THE INVENTION
[0006] The foregoing provides a non-exclusive list of the objectives achieved by embodiments the present invention.
[0007] It is an object of embodiments of the invention to reduce pulmonary viral titer and thereby treat, cure or prevent influenza infection, especially Type A influenza infection, and more especially strain H1N1 influenza infection.
[0008] It is a further object of embodiments of the invention to reduce pulmonary viral titer and treat, cure, or prevent influenza infection by administering CC10 by the intravenous route, the inhaled route, or the intranasal route (according to PCT 2009), or by a combination of routes.
[0009] It is another object of embodiments of the invention to reduce viral titer and treat, cure, or prevent viral infection by administering CC10 by the intravenous route, the inhaled route, or the intranasal route, the oral route, the intravaginal route, or by a combination of routes. It is yet another object of embodiments of the invention to inhibit viral replication at the cellular level using CC10 or other members of the secretoglobin family.
SUMMARY OF THE INVENTION
[0010] These and other objects, features and advantages are achieved by embodiments of the invention as defined in the appended claims.
[0011] In particular, the present invention relates to human uteroglobin or recombinant human uteroglobin for use in a method of treatment for reducing the titer of influenza virus in the lung tissue of a patient. In certain aspects, human uteroglobin or recombinant human uteroglobin is administered.
[0012] These and other objects, features and advantages are also achieved by embodiments of the invention by administering uteroglobin (CC10) in a dosage range given at appropriate intervals or in one dose where a patient is diagnosed with an influenza infection by symptoms of fever, myalgia, and congestion, and/or by detection of influenza virus in patient samples (nasal lavages, blood or sputum samples) through culturing of the virus, immunological detection of the virus, and/or detection of the viral nucleic acid, using standard methods.
[0013] In certain aspects of the invention, uteroglobin (CC10) is administered intranasally in a dose divided about equally between each nostril in a range of 1.5 micrograms to 1.5 milligrams per kilogram of body weight per day, or in multiple doses which taken together achieve this dosage range on a daily basis to reduce pulmonary viral titer and treat, cure or prevent influenza infection.
[0014] In another aspect, uteroglobin (CC10) is administered intravenously in a dose of up to 10 milligrams per kilogram of body weight per day, or in multiple doses which taken together achieve this dosage range on a daily basis to treat, cure or prevent influenza infection.
[0015] In another aspect, a non-human CC10 protein is administered in a dosage range given at appropriate intervals or in one dose where a patient is diagnosed with a viral infection by symptoms characteristic of the particular virus, and/or by detection of virus in patient samples through culturing of the virus, immunological detection of the virus, and/or detection of the viral nucleic acid, using standard methods.
BRIEF DESCRIPTION OF THE DRAWINGS
[0016]
Fig. 1 is a bar graph depicting H1N1 viral load at 2 days in lungs of infected cotton rats treated with intranasal rhCC10. Viral titer is expressed as (x 107) TCID50/gram of tissue.
Fig. 2 is a bar graph depicting H1N1 viral load at 2 days in lungs of cotton rats treated with intraperitoneal injection of rhCC10. Viral titer is expressed as (x 107) TCID50/gram of tissue.
Fig. 3 is a bar graph depicting inhibition of viral replication in cultured cells by rhCC10. RHCC10 was added to the culture medium of HEp2 cells at 100 micrograms/ml, 300 micrograms/ml, and 1 milli-gram/ml and left for 4 hours. Then the medium was removed and replaced and cells were infected with RSV for 1 hour. The cells were then washed to remove excess virus and rhCC10 was added back and incubated for another hour. Then cells were washed to remove excess CC10. Viral titers in culture medium were measured at 4 days post-infection. Each CC10 concentration was performed in triplicate.
Fig. 4 is a bar graph comparison of rhCC10 antiviral effects when given pre-infection and post-infection. HEp2 cells were treated with 1 mg/ml rhCC10 and infected with RSV as in figure 3. In addition, 1 mg/ml of rhCC10 was given at one hour after infection (treatment DO), 24 hours after infection (treatment D1 ), and 48 hours after infection (treatment D2). Viral titers in culture media were measured on day 4 postinfection.
DETAILED DESCRIPTION
[0017] Embodiments of the present invention relate to the use of CC10 to reduce pulmonary viral titer and treat, cure or prevent influenza infection. The CC10 is preferably a recombinant human CC10 protein (rhCC10) obtained by the processes described in U.S. Patent App. Publication No.: 20030207795 issued as US Pat. No. 7,122,344 and PCT/US09/43613 published as W02009/140269A2, or via any other process which yields pharmaceutical grade rhCC10. The rhCC10 of the embodiments of the present invention may be administered with, without, before or after other intranasal, pulmonary, or systemic therapy.
[0018] Without limiting the scope of possible synthetic processes that may be used to make human CC10, the recombinant human CC10 (aka uteroglobin) that is active in suppressing viral replication in vitro and in vivo was synthesized and characterized as described in US Patent App. Publication No.: 20030207795 issued as US Pat. No. 7,122,344.
[0019] Preparations of rhCC10 for intranasal administration as described in PCT/US09/43613 published as W02009/140269A2 represent further embodiments of the present invention that may be used to suppress viral replication in vivo, particularly in the nasal passages and sinuses.
Dosages [0020] Preferably, in treating or preventing influenza infection, rhCC10 is administered intranasally, to each nostril 1-3 times per day, for 7-14 days, and every other day thereafter for another 14 days, and thereafter as needed. More preferably, rhCC10 is administered as soon as the patient begins to experience fever, myalgia, and congestion or is diagnosed with influenza.
[0021] The rhCC10 may be produced in a process of producing a pharmaceutical grade recombinant human uteroglobin as described in paragraph 31 of US Pat. No. 7,122,344, which comprises the steps of: a) providing a bacterial expression system capable of expressing rhCC10; b) inoculating a fermenter with an inoculum comprising the bacterial expression system to form a fermentation culture; c) adding an induction agent to the fermentation culture to induce the expression of rhCC10 by the bacterial expression system; d) harvesting the rhCC10 expressed in step c; and e) purifying the rhCC10 harvested in step d, wherein the purifying step comprises the use of at least one filter and at least one ion exchange column. The rhCC10 may also be expressed in alternative bacterial, fungal, insect, mammalian, or plant expression systems and purified to meet specifications for a pharmaceutical product suitable for administration to humans using standard methods.
[0022] Specifications and testing results for pharmaceutical grade rhCC10, according to Table 20 of US Patent App. Publication No.: 20030207795 issued as US Pat. No. 7,122,344, that may be used to reduce viral titers include the following:
[0023] In a further embodiment, the rhCC10 of the present invention that inhibits viral replication also inhibits phospholipase A2 (PLA2) enzymes, as described in US Patent App. Publication No.: 20030207795, issued as US Pat. No. 7,122,344.
To effectuate the desired outcomes which are further described below, reference is made to methods of admin- { istration described in the following embodiments.
[0024] In one embodiment, a dose or multiple doses of intranasal rhCC10 equaling a dose ranging from about 1.5 micrograms to about 5 milligrams per kilogram of body weight per day may be administered. In another embodiment, rhCC10 may be administered in the dose range on a daily basis. In yet another embodiment, rhCC10 may be administered in the dose range on a daily basis for at least seven days consecutively. In still a further embodiment, rhCC10 may be administered in the dose range on a daily basis for at least 14 days consecutively. In still another embodiment, rhCC10 may be administered in the dose range every other day for 30 days consecutively. In yet another embodiment, rhCC10 may be administered in tapered dosages daily fortén consecutive days, said tapered dosages comprising a high dose at each administration for the first three days, an intermediate dose at each administration for the second three days, and a low dose at each administration for the last four days. In yet still another embodiment, rhCC10 may | be administered in the dose range or in tapered doses up to three times per day, approximately every eight hours.
[0025] In another embodiment, the above doses of rhCC10 may be administered intranasally to the patient as an aerosol, by intranasal spray or lavage, or by deposition of a gel or cream, or other method of instillation in the nasal passages.
[0026] In another embodiment, the above doses of rhCC10 may be administered by inhalation to the patient as an aerosol, by nebulizer or metered dose inhaler, or other method of direct application to the lungs and airways.
[0027] In another embodiment, in treating or preventing influenza infection, rhCC10 is administered intravenously, in doses of 15 micrograms to 20 milligrams per kilogram of body weight, 1 -3 times per day, for 7-14 days, and every other day thereafter for another 14 days, and thereafter as needed. In yet another embodiment, rhCCIO may be administered in tapered dosages daily fortén consecutive days, said tapered dosages comprising a high dose at each administration for the first three days, an intermediate dose at each administration for the second three days, and a low dose at each administration for the last four days. In yet still another embodiment, rhCCIO may be administered in the dose range or in tapered doses up to three times per day, approximately every eight hours.
[0028] In another embodiment the above doses of rhCC10 may be administered to the patient using a combination of intranasal, inhaled, and intravenous routes. In a further embodiment, rhCC10, in accordance with the methods described above, may be administered prior to, during or after anti-viral therapy, anti-biotic therapy, decongestant, anti-histamine, mucolytic, expectorant, mucus suppressor, surfactant, bronchodilator, vasoconstrictor, sinus pain analgesic, or other typical therapy. In still another embodiment, rhCCIO, in accordance with the methods described above, may be administered to reduce pulmonary viral titer and treat, cure, or prevent influenza infection.
[0029] The doses of rhCCIO and application methods described above may be administered daily, more than once daily, three times daily, every other day or in a tapered fashion depending upon the severity of influenza infection being treated, the patient’s overall health, and whether underlying conditions are present. For example, the more severe the infection, the higher the amount of rhCCIO would be required to effectively treat it. It is understood that a physician would be able to monitor and adjust doses, formulations, and application methods as needed based on the patient’s symptoms and responses to therapy and within the parameters and dose ranges described in the embodiments of the present invention.
Formulations [0030] Intranasal formulations, devices, and methods by which rhCCIO may be administered intranasally have been described in PCT/US09/43613, published as W02009/140269A2, which was incorporated in its en-tiretyin the text of the present PCT application as originally filed. The intravenous formulation of rhCCIO consists of a 5.5 mg/ml solution in 0.9% saline and has been described in U.S. Patent App. Publication No.: 20030207795, issued as US Pat. No. 7,122,344, which was incorporated in its entirety in the text of the present PCT application as originally filed. EXAMPLE 1
Propagation and titer determination of influenza virus [0031] TheA/PR/8/34 Influenza A viral strain (H1N1 ), purchased from the American Type Culture Collection (Manasass, Virginia, USA) is prepared. Influenza virus is propagated in MDCK cells (ATCC catalog# CCL-34) by infecting 60% confluent cell monolayer (150 cm2 flasks) with flu virus at a multiplicity of infection (MOI) of 0.01. Three to four days later, when cytopathic effect is generalized and most of the cells have detached from the culture vessel, the cells and supernatants are harvested. Cells are removed by centrifugation (800g) and the supernatant filtered (0.45 μίτι) and centrifuged (18000g) for 2 hours at 4°C to pellet viruses. The viral pellet is resuspended in DMEM medium, aliquoted and stored at -150°C. Influenza virus titer is determined by applying 0.1 mL of serially diluted viral stocks to MDCK cell monolayers in a 96-well plate cultured in the presence of 0.1% bovine serum albumin and trypsin. Three days later, cytopathic effects were scored and the tissue culture infectious dose 50% (TCID50) is determined using Kärber’s method.
Processing of Lung Tissues for viral load analysis.
[0032] Sections of the left and right lobes from lungs of infected mice and cotton rats are aseptically removed, weighed and homogenized in 1 ml of DMEM medium for 45 seconds using a tissue tearor apparatus (mod-el#985-370, Biopspec Products Inc.) at a setting of 5. Homogenates are centrifuged at 3000g for 20 minutes. Clarified supernatants are collected, and stored frozen at -150°C until used.
Determination of Viral Titers.
[0033] Viral titers in amplified viral stocks and in lung homogenates were determined by serial dilution followed by plaque forming assay (PFA) or foci forming assay (FFA). Plaque forming units (PFU) and foci forming units (FFU) per milliliter of original sample were calculated prior to the start of the study. One set of influenza samples sufficient for carrying out PFA and FFA was stored and PFU and FFU were determined after the completion of the studies. Serial dilutions of cultured virus in clarified media (DMEM with 1% BSA) were prepared across 101 to 108 dilution range. Each dilution is evaluated by a plaque forming assay (PFA) and a foci forming assay (FFA). Culture titers typically yield 107 - 109 pfu/ml for influenza. EXAMPLE 2
Intranasal administration of rhCCIO to reduce pulmonary influenza virus titer [0034] The cotton rat (S. hispidus), a type of vole, is an animal model in which influenza replicates and generates a mild respiratory infection (Ottolini, 2005). The animals are infected by intranasal inoculation with influenza virus and pulmonary viral titers peak two days (about 48 hours) after inoculation. This model is used to screen for compounds that inhibit influenza replication in vivo.
[0035] Pathogen free cotton rats were purchased from Virion Systems, Inc. (Rockville, MD). A total of eighteen cotton rats (S.hispidus, 6-8 weeks old) were infected with Type A influenza (A/PR/8/34), strain H1N1, by intranasal inoculation using 107 TCID50 in 0.1 ml volume for each rat. Six animals received a placebo (0.9% NaCI), six animals received 0.5 mg/kg of rhCC10 and six animals received 5.0 mg/kg of rhCC10 by intranasal instillation 2 hours before viral inoculation. Animals were sacrificed on day 2 post-infection when viral titers are typically highest and viral load was determined in lung tissue. Fig.1 illustrates the reductions in viral titer in lung tissue that were observed in the both rhCC10 dose groups. Viral titer in lung is expressed as (x 107) TCID50/gram of tissue. EXAMPLE 3
Systemic administration of rhCC10 to reduce pulmonary influenza virus titer [0036] A total of eighteen cotton rats (S.hispidus, 6-8 weeks old) were infected with Type A influenza (A/PR/8/34), strain H1N1, by intranasal inoculation using 107 TCID50 in 0.1 ml volume for each rat. Six animals received a saline placebo, six animals received 0.5 mg/kg of rhCC10 and six animals received 5.0 mg/kg of rhCC10 by intraperitoneal injection (IP). The IP route results in significant amounts of circulating rhCC10 and simulates the intravenous route of administration in humans. Each animal received a total of six doses of either placebo or rhCC10 approximately every 12 hours, including two doses (morning and afternoon) on the day before infection, two doses on the day of infection, and two doses on the day after infection (3 doses before infection, 3 doses after infection). Animals were sacrificed on day 2 post-infection when viral titers are typically highest and viral load was determined in lung tissue. Fig. 2 illustrates the statistically significant reduction (p<0.01) in viral titer in lung tissue that was observed in the 5 mg/kg rhCC10 dose group, and the trend towards a lower viral titer in the 0.5 mg/kg dose group. Viral titer in lung is expressed as (x 107) TCID50/gram of tissue.
[0037] Based on the foregoing, rhCC10 has been found to reduce viral titer in a respiratory infection, indicating the use of rhCC10 to treat, cure and/or prevent influenza infection. Accordingly, embodiments of the present invention provide an intranasal, and intravenous, or a combination rhCC10 based therapy effective at treating, curing or preventing influenza infection. EXAMPLE 4 CCIO-mediated inhibition of viral replication at the cellular level [0038] HEp2 cells (ATCC, Manassas, VA) were used to propagate RSV, strain A-2 (Advanced Biotechnolo gies, Inc., Columbia, MD) and generate viral stocks. Cells were plated at 50,000 cells/well in 48 well plates and grown in MEM with 10% FBS to ~80% confluence. Cells were pre-treated with CC10 in 0.5 mL MEM for 4 hours. Medium was then changed and RSV infections were performed using 1 x 106 TCID50 per 100 mm TC dish for 1 hour. Non-adsorbed virus was removed by washing and 0. 5 ml of MEM with 2% FBS, 4 mM L-glutamine, and rhCC10 was added. Supernatants were collected on day 4 post infection and the virus titrated. Figure 3 shows that a concentration of 1 mg/mlCC10virtually eliminated RSV production, while 100 and 300 micrograms/ml showed a ~3-fold decrease.
[0039] CC10 also inhibited viral replication in cells when given at 1, 24, and 48 hours after infection. Fig. 4 shows that rhCC10 is effective at reducing viral titer not only when added before infection, but also when added after infection. This is the first report of a direct anti-viral activity of CC10 at the cellular level and illustrates the potential utility of rhCC10 as an anti-viral therapy for postexposure treatment. EXAMPLE 5 CC10 anti-viral mechanism of action [0040] The phenotype of airway epithelial cells in the CC10 knockout mouse illustrates that in the absence of CC10, the distribution of intracellular organelles is abnormal, that abnormal stacked membranous structures are present, and that secretion of other proteins made by the cell is disrupted. We surmise that this phenotype means that CC10 plays an active role in transport of secretory vesicles from the Golgi apparatus to the plasma membrane of the cell. CC10 also modulates the uptake and processing of antigens in antigen-presenting cells. We interpret these observations to mean that CC10 is an important factor in the transport of materials both out of and in to many types of cells. We therefore infer that CC10 inhibits viral replication by interfering with viral transport in the cell. Since all viruses rely upon cellular transport processes to invade the cell and replicate, CC10 can be expected to inhibit the replication of all viruses. Likewise, other secretoglobins, which share similar structure to CC10, can also be expected to inhibit viral replication at the cellular level. Similarly, peptides derived from CC10 and other secretoglobins that modulate cellular transport processes can also be expected to inhibit viral replication.
Claims 1. Human uteroglobin or recombinant human uteroglobin for use in a method of treatment for reducing the titer of influenza virus in the lung tissue of a patient. 2. Human uteroglobin or recombinant human uteroglobin for use according to claim 1, wherein the human uteroglobin or recombinant human uteroglobin treats, cures, or prevents an influenza infection in the patient. 3. Human uteroglobin or recombinant human uteroglobin for use according to claim 1, wherein the influenza is Type A influenza. 4. Human uteroglobin or recombinant human uteroglobin for use according to claim 1, wherein the influenza is the H1N1 strain of influenza. 5. Human uteroglobin or recombinant human uteroglobin for use according to claim 1, wherein the human uteroglobin or recombinant human uteroglobin is administered by the intranasal route, the intravenous route or a combination of intranasal and intravenous routes. 6. Human uteroglobin or recombinant human uteroglobin for use according to claim 1, wherein the human uteroglobin or recombinant human uteroglobin inhibits viral replication at the cellular level. 7. Human uteroglobin or recombinant human uteroglobin for use according to claim 6, wherein the human uteroglobin or recombinant human uteroglobin is administered to an infected patient. 8. Human uteroglobin or recombinant human uteroglobin for use according to claim 6, wherein the human uteroglobin or recombinant human uteroglobin is administered to an infected animal. 9. Human uteroglobin or recombinant human uteroglobin for use according to claim 6, wherein the human uteroglobin or recombinant human uteroglobin is administered to an infected cell.
Patentansprüche 1. Menschliches Uteroglobin oder rekombinantes menschliches Uteroglobin zur Verwendung in einem Behandlungsverfahren zur Verminderung des Titers von Influenzavirus in dem Lungengewebe eines Patienten. 2. Menschliches Uteroglobin oder rekombinantes menschliches Uteroglobin zur Verwendung nach Anspruch 1, wobei das menschliche Uteroglobin oder das rekombinante menschliche Uteroglobin eine Influenzainfektion in dem Patienten behandelt, heilt oder verhindert. 3. Menschliches Uteroglobin oder rekombinantes menschliches Uteroglobin zur Verwendung nach Anspruch 1, wobei die Influenza Typ A-Influenza ist. 4. Menschliches Uteroglobin oder rekombinantes menschliches Uteroglobin zur Verwendung nach Anspruch 1, wobei die Influenza der H1N1-Stamm von Influenza ist. 5. Menschliches Uteroglobin oder rekombinantes menschliches Uteroglobin zur Verwendung nach Anspruch 1, wobei das menschliche Uteroglobin oder das rekombinante menschliche Uteroglobin durch den intranasalen Weg, den intravenösen Weg oder eine Kombination von intranasalen und intravenösen Wegen verabreicht wird. 6. Menschliches Uteroglobin oder rekombinantes menschliches Uteroglobin zur Verwendung nach Anspruch 1, wobei das menschliche Uteroglobin oderdas rekombinante menschliche Uteroglobin die virale Replikation auf der zellulären Ebene hemmt. 7. Menschliches Uteroglobin oder rekombinantes menschliches Uteroglobin zur Verwendung nach Anspruch 6, wobei das menschliche Uteroglobin oderdas rekombinante menschliche Uteroglobin an einen infizierten Patienten verabreicht wird. 8. Menschliches Uteroglobin oder rekombinantes menschliches Uteroglobin zur Verwendung nach Anspruch 6, wobei das menschliche Uteroglobin oderdas rekombinante menschliche Uteroglobin an ein infiziertes Tier verabreicht wird. 9. Menschliches Uteroglobin oder rekombinantes menschliches Uteroglobin zur Verwendung nach Anspruch 6, wobei das menschliche Uteroglobin oderdas rekombinante menschliche Uteroglobin an eine infizierte Zelle verabreicht wird.
Revendications 1. Utéroglobine humaine ou utéroglobine humaine recombinante pour une utilisation dans un procédé de traitement pour la réduction du titre du virus grippal dans le tissu pulmonaire d’un patient. 2. Utéroglobine humaine ou utéroglobine humaine recombinante pour une utilisation selon la revendication 1, dans laquelle l’utéroglobine humaine ou l’uté-roglobine humaine recombinante traite, guérit, ou prévient une infection grippale chez le patient. 3. Utéroglobine humaine ou utéroglobine humaine recombinante pour une utilisation selon la revendication 1, dans laquelle la grippe est la grippe de type A. 4. Utéroglobine humaine ou utéroglobine humaine recombinante pour une utilisation selon la revendication 1, dans laquelle la grippe est la souche H1 N1 de la grippe. 5. Utéroglobine humaine ou utéroglobine humaine recombinante pour une utilisation selon la revendication 1, dans laquelle l’utéroglobine humaine ou l’uté-roglobine humaine recombinante est administrée par la voie intranasale, la voie intraveineuse ou une combinaison des voies intranasale et intraveineuse. 6. Utéroglobine humaine ou utéroglobine humaine recombinante pour une utilisation selon la revendication 1, dans laquelle l’utéroglobine humaine ou l’utéroglobine humaine recombinante inhibe la réplication virale au niveau cellulaire. 7. Utéroglobine humaine ou utéroglobine humaine recombinante pour une utilisation selon la revendication 6, dans laquelle l’utéroglobine humaine ou l’utéroglobine humaine recombinante est administrée à un patient infecté. 8. Utéroglobine humaine ou utéroglobine humaine recombinante pour une utilisation selon la revendication 6, dans laquelle l’utéroglobine humaine ou l’utéroglobine humaine recombinante est administrée à un animal infecté. 9. Utéroglobine humaine ou utéroglobine humaine recombinante pour une utilisation selon la revendication 6, dans laquelle l’utéroglobine humaine ou l’utéroglobine humaine recombinante est administrée à une cellule infectée.
REFERENCES CITED IN THE DESCRIPTION
This list of references cited by the applicant is for the reader’s convenience only. It does not form part of the European patent document. Even though great care has been taken in compiling the references, errors or omissions cannot be excluded and the EPO disclaims all liability in this regard.
Patent documents cited in the description • US 61252028 PCT [0001] • US 20030207795 A [0017] [0018] [0022] [0023] [0030] • US 7122344 B [0017] [0018] [0021] [0022] [0023] • U S 0943613 W [0017] [0019] [0030] • WO 2009140269 A2 [0017] [0019] [0030] • US 7122344 PCT [0030]

Claims (2)

Szabadalmi igénypontok L3iâmàÂ^^^^^^’?etoÂÉ»I^SÂ^«rôj|l<^*n páciens Ä ,üf iliiiÄettilÄ^ »Igáié tpriebiö történő alkalmazásra. l. Humán uteroglobin vagy reknmbinüus humán uteroglobin sz I .igénypont .szerinti, alkalma·:. tàsra. ahol a humán lUeroglobin vagy r manó bináris humán uteroglobin keceli, gyógyítja mgy negolozt az bnluenza fertőzést a p;k nősben. 1 Humán uteroglobin vagy rekombtnáns humán uteroglobin âz I igénypont szerinti aikaingö msra, ahol tnt Inûuenaa A típusú inílnensa. I Ihmx .tseuvloou's,ms rykoniO nmo bonutt ittcrogioHr -> 1 s^oypom szenna ,ők thra-rnsra, ahol az influenza az influenza Hl NI törzs. S 11um*» uieu.giobtn vagy tekoutbmaoá humán tunogiohm ..« I. igénypont mrinfi alkalma-zásra.. ahol a hunján uterogtobioí vagy rekomhuians human uteroglobint imranazáltMtt, intravénásán y lg y az iutranazális és intravénás módszer kgmhmáck|ácal adagoljuk, \ human meo'ghdxn vagy teh-mbiuatís humfttt uteroglobin az ! > Igénypont ez^Hntí alkalmai zásra, ahol a hu inán uteroglobin sae.y rekombináns humán uteMglobiua sejt szintién gátolja a s Ints repiikációját.Claims L3iâmà ^^^^^^ '? EtoÂÉ »I ^ S ^ rôj | l <^ * n for use in the treatment of patients. l. Human uteroglobin or recombinant human uteroglobin I claim. TASR. where human leroglobin or rNA binary human uteroglobin is elicited, heals neglected bnluenza infection in p. k. A timing ms for human uteroglobin or recombinant human uteroglobin as claimed in claim 1, wherein t In Inuea is a type A inhibitor. Ihmx .tseuvloou's, ms rykoniO nmo bonutt herecrogioHr -> 1 s ^ oypom senna, they are thra-rns, where the flu is the influenza H1 NI strain. S11um * »uieu.giobtn or tekoutbmaoá human tunogroup. <RTI ID = 0.0> I. </RTI> mrinfi .. wherein the uterogenibi or rekomhuians human uteroglobin is imaged in the honey, administered intravenously by the iutranazal and intravenous method with kgmhm. 'ghdxn or teh-mbiuatís humfttt uteroglobin! > This is an application for this application, wherein the recombinant human uteMglobiua cell of the inan uteroglobin sae.y inhibits s ints replication. 7. Mámáó uteroglobin árggy: rékoiBbmábs inmin tiérogloÉna h,:i|enypmt szerinti alkalmin zásra. ahol a humán oieroglohmt vágy vekomhfnáns: humán spiúroglolmt egy ílrtőzétt pici a unú t- k a d a g<. d j uk. t. iekonibinän$f|uÄÄilltcroglobht a;C i|éb|gÖdt szeriqfl âlWlhif· gâ vagy rekombíháná human utirp|iöhmt eg| iemkbihÉf libák: adagoljuk. i, Myiaâfi ütétöf l!#|n :¥âgÿ- tíí^gf^ir* a 6·. lœrmtl a! ka Ima- aásria ahol a hooma üiorogfobmt vagy lekarnhlMM hoffiàa iftarogioMot agy Ihitéaott sejihez· adpk..7. Relaxing uteroglobin argyg: rékoiBbmábs inmin térogloÉna h,: i | enypmt. wherein the human oieroglohmt desire is vecchymnant: human spuroglycol is an exaggerated smallpox. d j uk. t. sisonibinän $ f | uÄÄilltcroglobht a; c i | éb | gödl seriqfl llWlhif · gâ or recombinant human utirp | iöhmt eg | downy geese: added. i, Myiath's lf # | n: ¥ âgÿtíii gg ^ and * a 6 ·. lœrmtl a! ka-ria ria ahol ria ria ria ria ria ria ria ria ahol ahol ahol ahol ahol ahol ahol ahol ahol ahol ahol ahol ahol ahol ahol ahol ahol ahol ahol ahol ahol ahol ahol
HUE10824032A 2009-10-15 2010-10-13 Recombinant human cc10 protein for treatment of influenza HUE030838T2 (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US25202809P 2009-10-15 2009-10-15

Publications (1)

Publication Number Publication Date
HUE030838T2 true HUE030838T2 (en) 2017-06-28

Family

ID=43876512

Family Applications (1)

Application Number Title Priority Date Filing Date
HUE10824032A HUE030838T2 (en) 2009-10-15 2010-10-13 Recombinant human cc10 protein for treatment of influenza

Country Status (18)

Country Link
US (1) US8957018B2 (en)
EP (1) EP2488205B1 (en)
JP (2) JP5944316B2 (en)
KR (1) KR101597391B1 (en)
CN (1) CN102834115B (en)
AU (1) AU2010306840B2 (en)
BR (1) BR112012008463A2 (en)
CA (1) CA2813740C (en)
DK (1) DK2488205T3 (en)
ES (1) ES2606541T3 (en)
HU (1) HUE030838T2 (en)
IL (2) IL219009A0 (en)
MX (1) MX2012004409A (en)
NZ (1) NZ599511A (en)
PL (1) PL2488205T3 (en)
PT (1) PT2488205T (en)
RU (1) RU2554745C2 (en)
WO (1) WO2011047065A1 (en)

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060281681A1 (en) 1997-05-28 2006-12-14 Pilon Aprile L Methods and compositions for the reduction of neutrophil influx and for the treatment of bronchpulmonary dysplasia, respiratory distress syndrome, chronic lung disease, pulmonary fibrosis, asthma and chronic obstructive pulmonary disease
EP2303308A4 (en) 2008-05-13 2012-11-07 Clarassance Inc Recombinant human cc10 and compositions thereof for use in the treatment of nasal rhinitis
US9168285B2 (en) 2009-10-15 2015-10-27 Therabron Therapeutics, Inc. Recombinant human CC10 protein for treatment of influenza and ebola
NZ599511A (en) * 2009-10-15 2014-06-27 Clarassance Inc Recombinant human cc10 protein for treatment of influenza
US9394349B2 (en) * 2013-03-15 2016-07-19 Therabron Therapeutics, Inc. Modification and compositions of human secretoglobin proteins
US20140274915A1 (en) 2013-03-15 2014-09-18 Clarassance, Inc. Methods of Use for Recombinant Human Secretoglobins
US11690893B2 (en) * 2018-11-30 2023-07-04 Apc Research Assets Llc Compositions and methods of use for secretoglobins to protect the glycocalyx via interactions with heparan sulfate proteoglycan proteins
US20230241163A1 (en) * 2020-06-02 2023-08-03 Apc Research Assets Llc Recombinant Human CC10 Protein for Treatment of Influenza, Ebola and Coronavirus

Family Cites Families (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4691009A (en) 1984-12-26 1987-09-01 Repligen Corporation Hybrid proteins produced by an ultrahigh prokaryotic expression system
US4917826A (en) 1985-10-18 1990-04-17 The Upjohn Company Cyclic hydrocarbons with an aminoalkyl sidechain
US4820514A (en) 1985-12-30 1989-04-11 Texas A&M University System Low dosage of interferon to enhance vaccine efficiency
JP2656944B2 (en) 1987-04-30 1997-09-24 クーパー ラボラトリーズ Aerosolization of protein therapeutics
US5266562A (en) 1987-11-19 1993-11-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Anti-inflammatory agents
US5354269A (en) 1991-12-20 1994-10-11 Fibrogenex, Inc. Method for treating cancer resections
US5491130A (en) 1992-11-10 1996-02-13 The United States Of America As Represented By The Department Of Health And Human Services Peptide inhibitors of fibronectin and related collagen-binding proteins
US5482930A (en) 1993-06-09 1996-01-09 The Regents Of The University Of California Anti-inflammatory composition and method with des-Tyr dynorphin and analogues
US5470885A (en) 1993-09-29 1995-11-28 The Research Foundation Of The State University Of New York Fluorocarbons as anti-inflammatory agents
FR2724665B1 (en) 1994-09-16 1996-12-20 Rhone Poulenc Rorer Sa PROCESS FOR PRODUCING RECOMBINANT PROTEINS, PLASMIDS AND MODIFIED CELLS
US5935860A (en) 1995-03-07 1999-08-10 The George Washington University Use of uteroglobin expression as a molecular marker for prostatic intraepithelial neoplasia
US5696092A (en) 1995-03-07 1997-12-09 George Washington University Methods and compositions for inhibiting metastasis of epithelial cell-derived cancers
AU6290496A (en) 1995-06-27 1997-01-30 Igen International, Inc. High-level expression and efficient recovery of ubiquitin fusion proteins from escherichia coli
US5817750A (en) 1995-08-28 1998-10-06 La Jolla Cancer Research Foundation Structural mimics of RGD-binding sites
CA2248136A1 (en) 1996-03-21 1997-09-25 Human Genome Sciences, Inc. Human endometrial specific steroid-binding factor i, ii and iii
US6110744A (en) * 1996-11-13 2000-08-29 Board Of Regents, The University Of Texas System Diminishing viral gene expression by promoter replacement
US20050261180A1 (en) 1997-05-28 2005-11-24 Pilon Aprile L Use of recombinant human uteroglobin in treatment of inflammatory and fibrotic conditions
US6255281B1 (en) 1997-05-28 2001-07-03 Claragen, Inc. And U.S. Government Use of recombinant human uteroglobin in treatment of inflammatory and fibrotic conditions
US20030207795A1 (en) 1997-05-28 2003-11-06 Pilon Aprile L. Methods for the production of purified recombinant human uteroglobin for the treatment of inflammatory and fibrotic conditions
US20020160948A1 (en) 1998-07-21 2002-10-31 Aprile Pilon Recombinant human uteroglobin in treatment of inflammatory and fibrotic conditions
US20020169108A1 (en) 1997-05-28 2002-11-14 Pilon Aprile L. Methods and compositions for the treatment of fibrotic conditions & impaired lung function & to enhance lymphocyte production
US20060025348A1 (en) 1997-05-28 2006-02-02 Pilon Aprile L Methods and compositions for the treatment of fibrotic conditions & impaired lung function & to enhance lymphocyte production
US20040047857A1 (en) 1997-05-28 2004-03-11 Pilon Aprile L. Methods and compositions for the treatment of fibrotic conditions & impaired lung function & to enhance lymphocyte production
US7122344B2 (en) 1997-05-28 2006-10-17 Claragen, Inc. Methods for the production of purified recombinant human uteroglobin for the treatment of inflammatory and fibrotic conditions
US20060281681A1 (en) 1997-05-28 2006-12-14 Pilon Aprile L Methods and compositions for the reduction of neutrophil influx and for the treatment of bronchpulmonary dysplasia, respiratory distress syndrome, chronic lung disease, pulmonary fibrosis, asthma and chronic obstructive pulmonary disease
US20030008816A1 (en) 1997-05-28 2003-01-09 Pilon Aprile L. Methods and compositions for the treatment of fibrotic conditions & impaired lung function & to enhance lymphocyte production
BR9909626A (en) 1998-04-16 2002-01-15 Texas Biotechnology Corp Compounds that inhibit integrin binding to its receptors
AU5836700A (en) * 1999-06-01 2000-12-18 Patrick T. Prendergast Peptides for therapeutic use
EP1235846A4 (en) * 1999-11-04 2004-07-14 Human Genome Sciences Inc Uteroglobin-like polynucleotides, polypeptides, and antibodies
CA2405946A1 (en) 2000-04-14 2001-10-25 Claragen, Inc. Methods and compositions for the treatment of fibrotic conditions and impaired lung function and to enhance lymphocyte production
AU2001282977A1 (en) 2000-07-26 2002-02-05 The Burnham Institute Screening methods based on superactivated alphavbeta3 integrin
WO2003057257A1 (en) 2002-01-02 2003-07-17 The Johns Hopkins University Cc10 inhibits th2 cytokines and eotaxins involved in allergic diseases
US20090227025A1 (en) * 2003-06-06 2009-09-10 The Board Of Regents Of The University Of Texas System Ex vivo human lung/immune system model using tissue engineering for studying microbial pathogens with lung tropism
US20090253174A1 (en) * 2008-04-08 2009-10-08 Zach Serber Expression of Heterologous Sequences
EP2303308A4 (en) 2008-05-13 2012-11-07 Clarassance Inc Recombinant human cc10 and compositions thereof for use in the treatment of nasal rhinitis
NZ599511A (en) 2009-10-15 2014-06-27 Clarassance Inc Recombinant human cc10 protein for treatment of influenza

Also Published As

Publication number Publication date
EP2488205A4 (en) 2013-04-03
CN102834115A (en) 2012-12-19
PT2488205T (en) 2016-11-10
CN102834115B (en) 2015-01-14
RU2554745C2 (en) 2015-06-27
PL2488205T3 (en) 2018-01-31
BR112012008463A2 (en) 2019-09-24
IL219009A0 (en) 2012-06-28
AU2010306840A1 (en) 2012-05-17
JP2013508283A (en) 2013-03-07
AU2010306840B2 (en) 2015-06-25
IL243697A0 (en) 2016-04-21
CA2813740A1 (en) 2011-04-21
RU2012120671A (en) 2013-11-20
KR101597391B1 (en) 2016-02-24
CA2813740C (en) 2016-08-02
ES2606541T3 (en) 2017-03-24
EP2488205A1 (en) 2012-08-22
EP2488205B1 (en) 2016-09-21
WO2011047065A1 (en) 2011-04-21
JP5944316B2 (en) 2016-07-05
MX2012004409A (en) 2012-08-23
US20120231997A1 (en) 2012-09-13
KR20120098699A (en) 2012-09-05
US8957018B2 (en) 2015-02-17
JP2016155799A (en) 2016-09-01
NZ599511A (en) 2014-06-27
DK2488205T3 (en) 2017-01-16

Similar Documents

Publication Publication Date Title
DK2488205T3 (en) Recombinant human CC10 protein for the treatment of influenza
CN104395336B (en) With the extensive reactive antigen of the H5N1 and H1N1 influenza viruses of calculation optimization
RU2672888C1 (en) Anti-viral immunotropic agent for treatment of arvi
RU2524304C2 (en) Application of acetylsalicylic acid salt for treatment of viral infections
JPH06507404A (en) How to treat infectious respiratory diseases
Li et al. Intranasal co-administration of 1, 8-cineole with influenza vaccine provide cross-protection against influenza virus infection
JP2014501718A (en) Composition comprising peptide and viral neuraminidase inhibitor
CN112912074A (en) EGCG-palmitate compositions and methods of use thereof
US20230241163A1 (en) Recombinant Human CC10 Protein for Treatment of Influenza, Ebola and Coronavirus
US9168285B2 (en) Recombinant human CC10 protein for treatment of influenza and ebola
US20220193021A1 (en) Combination therapy for treating influenza virus infection
KR20230021009A (en) Azelastine as an antiviral treatment
WO2016133560A1 (en) Recombinant human cc10 protein for treatment of influenza and ebola
US20110091484A1 (en) Compositions and methods for preventing or treating a viral infection
US20240108713A1 (en) Novel replication deficient influenza a virus inducing high levels of type i interferon
WO2024096743A1 (en) Sars-cov-2 binding antibody
SE545271C2 (en) Pharmaceutical composition for treatment of viral infections caused by enveloped viruses
WO2022133455A1 (en) Diphenhydramine and lactoferrin for prevention and treatment of covid-19
WO2024096742A1 (en) SARS-CoV-2 BINDING POLYPEPTIDE
WO2021207653A1 (en) Use of a tlr9 agonist in methods for treating covid-19
CN116635058A (en) Antiviral agent
EP1596851A1 (en) The use of sodium 2-mercaptoethanesulfonate as antiviral agent
EP3886902A1 (en) Methods and compositions for preventing or treating acute exacerbations with polyclonal immunoglobulin
Ramasamy Immunity to human influenza A—An overview
Sankovski et al. Bovine Colostrum Derived Antibodies Against SARS-CoV-2 Show Great Potential to Serve as a Prophylactic Agent